Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Infect Immun ; 91(11): e0024023, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37847031

RESUMO

Cystic fibrosis (CF) is a human genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene that encodes a chloride channel. The most severe clinical manifestation is associated with chronic pulmonary infections by pathogenic and opportunistic microbes. Drosophila melanogaster has become the invertebrate model of choice for modeling microbial infections and studying the induced innate immune response. Here, we review its contribution to the understanding of infections with six major pathogens associated with CF (Staphylococcus aureus, Pseudomonas aeruginosa, Burkholderia cepacia, Mycobacterium abscessus, Streptococcus pneumoniae, and Aspergillus fumigatus) together with the perspectives opened by the recent availability of two CF models in this model organism.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Animais , Humanos , Fibrose Cística/microbiologia , Drosophila melanogaster , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Pulmão/microbiologia , Aspergillus fumigatus , Imunidade Inata , Pseudomonas aeruginosa
2.
Microbiol Spectr ; 11(4): e0077723, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37260399

RESUMO

Mycobacterium abscessus, an intracellular nontuberculous mycobacterium, is considered the most pathogenic species among the group of rapidly growing mycobacteria. The resistance of M. abscessus to the host innate response contributes to its pathogenicity in addition to several virulence factors. We have recently shown in Drosophila that antimicrobial peptides (AMPs), whose production is induced by M. abscessus, are unable to control mycobacterial infection. This could be due to their inability to kill mycobacteria and/or the hidden location of the pathogen in phagocytic cells. Here, we demonstrate that the rapid internalization of M. abscessus by Drosophila macrophages allows it to escape the AMP-mediated humoral response. By depleting phagocytes in AMP-deficient flies, we found that several AMPs were required for the control of extracellular M. abscessus. This was confirmed in the Tep4 opsonin-deficient flies, which we show can better control M. abscessus growth and have increased survival through overproduction of some AMPs, including Defensin. Furthermore, Defensin alone was sufficient to kill extracellular M. abscessus both in vitro and in vivo and control its infection. Collectively, our data support that Tep4-mediated opsonization of M. abscessus allows its escape and resistance toward the Defensin bactericidal action in Drosophila. IMPORTANCE Mycobacterium abscessus, an opportunistic pathogen in cystic fibrosis patients, is the most pathogenic species among the fast-growing mycobacteria. How M. abscessus resists the host innate response before establishing an infection remains unclear. Using Drosophila, we have recently demonstrated that M. abscessus resists the host innate response by surviving the cytotoxic lysis of the infected phagocytes and the induced antimicrobial peptides (AMPs), including Defensin. In this work, we demonstrate that M. abscessus resists the latter response by being rapidly internalized by Drosophila phagocytes. Indeed, by combining in vivo and in vitro approaches, we show that Defensin is able to control extracellular M. abscessus infection through a direct bactericidal action. In conclusion, we report that M. abscessus escapes the host AMP-mediated humoral response by taking advantage of its internalization by the phagocytes.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium , Animais , Drosophila , Opsonização , Peptídeos Antimicrobianos , Defensinas/farmacologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Antibacterianos/farmacologia
3.
PLoS Pathog ; 19(3): e1011257, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36972320

RESUMO

Mycobacterium abscessus is the most pathogenic species among the predominantly saprophytic fast-growing mycobacteria. This opportunistic human pathogen causes severe infections that are difficult to eradicate. Its ability to survive within the host was described mainly with the rough (R) form of M. abscessus, which is lethal in several animal models. This R form is not present at the very beginning of the disease but appears during the progression and the exacerbation of the mycobacterial infection, by transition from a smooth (S) form. However, we do not know how the S form of M. abscessus colonizes and infects the host to then multiply and cause the disease. In this work, we were able to show the hypersensitivity of fruit flies, Drosophila melanogaster, to intrathoracic infections by the S and R forms of M. abscessus. This allowed us to unravel how the S form resists the innate immune response developed by the fly, both the antimicrobial peptides- and cellular-dependent immune responses. We demonstrate that intracellular M. abscessus was not killed within the infected phagocytic cells, by resisting lysis and caspase-dependent apoptotic cell death of Drosophila infected phagocytes. In mice, in a similar manner, intra-macrophage M. abscessus was not killed when M. abscessus-infected macrophages were lysed by autologous natural killer cells. These results demonstrate the propensity of the S form of M. abscessus to resist the host's innate responses to colonize and multiply within the host.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Infecções por Mycobacterium , Mycobacterium abscessus , Mycobacterium , Animais , Humanos , Camundongos , Drosophila melanogaster , Fagócitos/patologia , Infecções por Mycobacterium/microbiologia , Drosophila , Infecções por Mycobacterium não Tuberculosas/microbiologia
4.
J Antimicrob Chemother ; 77(12): 3496-3503, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36253948

RESUMO

BACKGROUND: Mycobacterium abscessus (Mabs), a rapidly growing Mycobacterium species, is considered an MDR organism. Among the standard antimicrobial multi-drug regimens against Mabs, amikacin is considered as one of the most effective. Parenteral amikacin, as a consequence of its inability to penetrate inside the cells, is only active against extracellular mycobacteria. The use of inhaled liposomal amikacin may yield improved intracellular efficacy by targeting Mabs inside the cells, while reducing its systemic toxicity. OBJECTIVES: To evaluate the colocalization of an amikacin liposomal inhalation suspension (ALIS) with intracellular Mabs, and then to measure its intracellular anti-Mabs activity. METHODS: We evaluated the colocalization of ALIS with Mabs in eukaryotic cells such as macrophages (THP-1 and J774.2) or pulmonary epithelial cells (BCi-NS1.1 and MucilAir), using a fluorescent ALIS and GFP-expressing Mabs, to test whether ALIS reaches intracellular Mabs. We then evaluated the intracellular anti-Mabs activity of ALIS inside macrophages using cfu and/or luminescence. RESULTS: Using confocal microscopy, we demonstrated fluorescent ALIS and GFP-Mabs colocalization in macrophages and epithelial cells. We also showed that ALIS was active against intracellular Mabs at a concentration of 32 to 64 mg/L, at 3 and 5 days post-infection. Finally, ALIS intracellular activity was confirmed when tested against 53 clinical Mabs isolates, showing intracellular growth reduction for nearly 80% of the isolates. CONCLUSIONS: Our experiments demonstrate the intracellular localization and intracellular contact between Mabs and ALIS, and antibacterial activity against intracellular Mabs, showing promise for its future use for Mabs pulmonary infections.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium , Humanos , Amicacina/farmacologia , Células Eucarióticas , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Lipossomos , Testes de Sensibilidade Microbiana
5.
PLoS Pathog ; 18(8): e1010771, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35960766

RESUMO

ESX type VII secretion systems are complex secretion machineries spanning across the mycobacterial membrane and play an important role in pathogenicity, nutrient uptake and conjugation. We previously reported the role of ESX-4 in modulating Mycobacterium abscessus intracellular survival. The loss of EccB4 was associated with limited secretion of two effector proteins belonging to the WXG-100 family, EsxU and EsxT, and encoded by the esx-4 locus. This prompted us to investigate the function of M. abscessus EsxU and EsxT in vitro and in vivo. Herein, we show that EsxU and EsxT are substrates of ESX-4 and form a stable 1:1 heterodimer that permeabilizes artificial membranes. While expression of esxU and esxT was up-regulated in M. abscessus-infected macrophages, their absence in an esxUT deletion mutant prevented phagosomal membrane disruption while maintaining M. abscessus in an unacidified phagosome. Unexpectedly, the esxUT deletion was associated with a hyper-virulent phenotype, characterised by increased bacterial loads and mortality in mouse and zebrafish infection models. Collectively, these results demonstrate that the presence of EsxU and EsxT dampens survival and persistence of M. abscessus during infection.


Assuntos
Mycobacterium abscessus , Mycobacterium marinum , Mycobacterium tuberculosis , Mycobacterium , Sistemas de Secreção Tipo VII , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Camundongos , Mycobacterium/genética , Mycobacterium abscessus/genética , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/genética , Sistemas de Secreção Tipo VII/genética , Sistemas de Secreção Tipo VII/metabolismo , Peixe-Zebra/metabolismo
6.
Microbiology (Reading) ; 167(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34224347

RESUMO

Non-tuberculous mycobacteria (NTM) are a large group of micro-organisms comprising more than 200 individual species. Most NTM are saprophytic organisms and are found mainly in terrestrial and aquatic environments. In recent years, NTM have been increasingly associated with infections in both immunocompetent and immunocompromised individuals, prompting significant efforts to understand the diverse pathogenic and signalling traits of these emerging pathogens. Since the discovery of Type VII secretion systems (T7SS), there have been significant developments regarding the role of these complex systems in mycobacteria. These specialised systems, also known as Early Antigenic Secretion (ESX) systems, are employed to secrete proteins across the inner membrane. They also play an essential role in virulence, nutrient uptake and conjugation. Our understanding of T7SS in mycobacteria has significantly benefited over the last few years, from the resolution of ESX-3 structure in Mycobacterium smegmatis, to ESX-5 structures in Mycobacterium xenopi and Mycobacterium tuberculosis. In addition, ESX-4, considered until recently as a non-functional system in both pathogenic and non-pathogenic mycobacteria, has been proposed to play an important role in the virulence of Mycobacterium abscessus; an increasingly recognized opportunistic NTM causing severe lung diseases. These major findings have led to important new insights into the functional mechanisms of these biological systems, their implication in virulence, nutrient acquisitions and cell wall shaping, and will be discussed in this review.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Mycobacterium não Tuberculosas/microbiologia , Micobactérias não Tuberculosas/metabolismo , Sistemas de Secreção Tipo VII/metabolismo , Proteínas de Bactérias/genética , Parede Celular/genética , Parede Celular/metabolismo , Humanos , Micobactérias não Tuberculosas/genética , Micobactérias não Tuberculosas/patogenicidade , Sistemas de Secreção Tipo VII/genética , Virulência
7.
PLoS Pathog ; 15(11): e1008069, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31703112

RESUMO

Free-living amoebae are thought to represent an environmental niche in which amoeba-resistant bacteria may evolve towards pathogenicity. To get more insights into factors playing a role for adaptation to intracellular life, we characterized the transcriptomic activities of the emerging pathogen Mycobacterium abscessus in amoeba and murine macrophages (Mϕ) and compared them with the intra-amoebal transcriptome of the closely related, but less pathogenic Mycobacterium chelonae. Data on up-regulated genes in amoeba point to proteins that allow M. abscessus to resist environmental stress and induce defense mechanisms, as well as showing a switch from carbohydrate carbon sources to fatty acid metabolism. For eleven of the most upregulated genes in amoeba and/or Mϕ, we generated individual gene knock-out M. abscessus mutant strains, from which ten were found to be attenuated in amoeba and/or Mϕ in subsequence virulence analyses. Moreover, transfer of two of these genes into the genome of M. chelonae increased the intra-Mϕ survival of the recombinant strain. One knock-out mutant that had the gene encoding Eis N-acetyl transferase protein (MAB_4532c) deleted, was particularly strongly attenuated in Mϕ. Taken together, M. abscessus intra-amoeba and intra-Mϕ transcriptomes revealed the capacity of M. abscessus to adapt to an intracellular lifestyle, with amoeba largely contributing to the enhancement of M. abscessus intra-Mϕ survival.


Assuntos
Amoeba/genética , Macrófagos/metabolismo , Infecções por Mycobacterium não Tuberculosas/genética , Mycobacterium abscessus/patogenicidade , Transcriptoma , Fatores de Virulência/genética , Virulência/genética , Amoeba/crescimento & desenvolvimento , Amoeba/microbiologia , Animais , Proteínas de Bactérias/genética , Macrófagos/microbiologia , Camundongos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/isolamento & purificação
9.
Proc Natl Acad Sci U S A ; 115(43): E10147-E10156, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30301802

RESUMO

Mycobacterium abscessus is a peculiar rapid-growing Mycobacterium (RGM) capable of surviving within eukaryotic cells thanks to an arsenal of virulence genes also found in slow-growing mycobacteria (SGM), such as Mycobacterium tuberculosis A screen based on the intracellular survival in amoebae and macrophages (MΦ) of an M. abscessus transposon mutant library revealed the important role of MAB_0855, a yet uncharacterized Mycobacterial membrane protein Large (MmpL). Large-scale comparisons with SGM and RGM genomes uncovered MmpL12 proteins as putative orthologs of MAB_0855 and a locus-scale synteny between the MAB_0855 and Mycobacterium chelonae mmpL8 loci. A KO mutant of the MAB_0855 gene, designated herein as mmpL8MAB , had impaired adhesion to MΦ and displayed a decreased intracellular viability. Despite retaining the ability to block phagosomal acidification, like the WT strain, the mmpL8MAB mutant was delayed in damaging the phagosomal membrane and in making contact with the cytosol. Virulence attenuation of the mutant was confirmed in vivo by impaired zebrafish killing and a diminished propensity to induce granuloma formation. The previously shown role of MmpL in lipid transport prompted us to investigate the potential lipid substrates of MmpL8MAB Systematic lipid analysis revealed that MmpL8MAB was required for the proper expression of a glycolipid entity, a glycosyl diacylated nonadecyl diol (GDND) alcohol comprising different combinations of oleic and stearic acids. This study shows the importance of MmpL8MAB in modifying interactions between the bacteria and phagocytic cells and in the production of a previously unknown glycolipid family.


Assuntos
Proteínas de Bactérias/metabolismo , Glicolipídeos/metabolismo , Mycobacterium abscessus/metabolismo , Fatores de Virulência/metabolismo , Virulência/fisiologia , Amoeba/microbiologia , Animais , Transporte Biológico/fisiologia , Linhagem Celular , Citosol/metabolismo , Humanos , Lipídeos , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Camundongos , Fagossomos/microbiologia , Peixe-Zebra/microbiologia
10.
J Vis Exp ; (139)2018 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-30320743

RESUMO

What differentiates Mycobacterium abscessus from other saprophytic mycobacteria is the ability to resist phagocytosis by human macrophages and the ability to multiply inside such cells. These virulence traits render M. abscessus pathogenic, especially in vulnerable hosts with underlying structural lung disease, such as cystic fibrosis, bronchiectasis or tuberculosis. How patients become infected with M. abscessus remains unclear. Unlike many mycobacteria, M. abscessus is not found in the environment but might reside inside amoebae, environmental phagocytes that represent a potential reservoir for M. abscessus. Indeed, M. abscessus is resistant to amoebal phagocytosis and the intra-amoeba life seems to increase M. abscessus virulence in an experimental model of infection. However, little is known about M. abscessus virulence in itself. To decipher the genes conferring an advantage to M. abscessus intracellular life, a screening of a M. abscessus transposon mutant library was developed. In parallel, a method of RNA extraction from intracellular Mycobacteria after co-culture with amoebae was developed. This method was validated and allowed the sequencing of whole M. abscessus transcriptomes inside the cells; providing, for the first time, a global view on M. abscessus adaptation to intracellular life. Both approaches give us an insight into M. abscessus virulence factors that enable M. abscessus to colonize the airways in humans.


Assuntos
Eucariotos , Mycobacterium abscessus/genética , Mycobacterium abscessus/patogenicidade , Fagócitos/microbiologia , Humanos , Virulência , Fatores de Virulência/genética
11.
Proc Natl Acad Sci U S A ; 115(5): E1002-E1011, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29343644

RESUMO

Mycobacterium abscessus, a rapidly growing mycobacterium (RGM) and an opportunistic human pathogen, is responsible for a wide spectrum of clinical manifestations ranging from pulmonary to skin and soft tissue infections. This intracellular organism can resist the bactericidal defense mechanisms of amoebae and macrophages, an ability that has not been observed in other RGM. M. abscessus can up-regulate several virulence factors during transient infection of amoebae, thereby becoming more virulent in subsequent respiratory infections in mice. Here, we sought to identify the M. abscessus genes required for replication within amoebae. To this end, we constructed and screened a transposon (Tn) insertion library of an M. abscessus subspecies massiliense clinical isolate for attenuated clones. This approach identified five genes within the ESX-4 locus, which in M. abscessus encodes an ESX-4 type VII secretion system that exceptionally also includes the ESX conserved EccE component. To confirm the screening results and to get further insight into the contribution of ESX-4 to M. abscessus growth and survival in amoebae and macrophages, we generated a deletion mutant of eccB4 that encodes a core structural element of ESX-4. This mutant was less efficient at blocking phagosomal acidification than its parental strain. Importantly, and in contrast to the wild-type strain, it also failed to damage phagosomes and showed reduced signs of phagosome-to-cytosol contact, as demonstrated by a combination of cellular and immunological assays. This study attributes an unexpected and genuine biological role to the underexplored mycobacterial ESX-4 system and its substrates.


Assuntos
Amoeba/microbiologia , Mycobacterium abscessus/patogenicidade , Fagossomos/microbiologia , Sistemas de Secreção Tipo IV/genética , Fatores de Virulência/genética , Proteínas de Bactérias/genética , Caspase 1/metabolismo , Cromatografia em Camada Delgada , Citosol/metabolismo , Ativação Enzimática , Citometria de Fluxo , Galectina 3/metabolismo , Deleção de Genes , Genômica , Humanos , Lipídeos/química , Macrófagos/microbiologia , Mutação , Mycobacterium abscessus/genética , Mycobacterium tuberculosis/patogenicidade , Células THP-1 , Virulência
12.
Mol Microbiol ; 104(6): 889-904, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28340510

RESUMO

Mycobacterial genomes contain large sets of loci encoding membrane proteins that belong to a family of multidrug resistance pumps designated Resistance-Nodulation-Cell Division (RND) permeases. Mycobacterial membrane protein Large (MmpL) transporters represent a subclass of RND transporters known to participate in the export of lipid components across the cell envelope. These surface-exposed lipids with unusual structures play key roles in the physiology of mycobacteria and/or can act as virulence factors and immunomodulators. Defining the substrate specificity of MmpLs and their mechanisms of regulation helps understanding how mycobacteria elaborate their complex cell wall. This review describes the diversity of MmpL proteins in mycobacteria, emphasising their high abundance in a few opportunistic rapid-growing mycobacteria. It reports the conservation of mmpL loci between Mycobacterium tuberculosis and non-tuberculous mycobacteria, useful in predicting the role of MmpLs with unknown functions. Paradoxically, whereas MmpLs participate in drug resistance mechanisms, they represent also attractive pharmacological targets, opening the way for exciting translational applications. The most recent advances regarding structural/functional information are also provided to explain the molecular basis underlying the proton-motive force driven lipid transport. Overall, this review emphasises the Janus-face nature of MmpLs at the crossroads between antibiotic resistance mechanisms and exquisite vulnerability to drugs.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Sequência de Aminoácidos , Anti-Infecciosos/metabolismo , Proteínas de Bactérias/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Parede Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Mycobacterium tuberculosis/metabolismo , Estrutura Terciária de Proteína , Força Próton-Motriz , Fatores de Virulência/metabolismo
13.
Open Biol ; 6(11)2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27906132

RESUMO

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium responsible for pulmonary and cutaneous infections in immunocompetent patients and in patients with Mendelian disorders, such as cystic fibrosis (CF). Mycobacterium abscessus is known to transition from a smooth (S) morphotype with cell surface-associated glycopeptidolipids (GPL) to a rough (R) morphotype lacking GPL. Herein, we show that M. abscessus S and R variants are able to grow inside macrophages and are present in morphologically distinct phagosomes. The S forms are found mostly as single bacteria within phagosomes characterized by a tightly apposed phagosomal membrane and the presence of an electron translucent zone (ETZ) surrounding the bacilli. By contrast, infection with the R form leads to phagosomes often containing more than two bacilli, surrounded by a loose phagosomal membrane and lacking the ETZ. In contrast to the R variant, the S variant is capable of restricting intraphagosomal acidification and induces less apoptosis and autophagy. Importantly, the membrane of phagosomes enclosing the S forms showed signs of alteration, such as breaks or partial degradation. Although not frequently encountered, these events suggest that the S form is capable of provoking phagosome-cytosol communication. In conclusion, M. abscessus S exhibits traits inside macrophages that are reminiscent of slow-growing mycobacterial species.


Assuntos
Macrófagos/microbiologia , Mycobacterium chelonae/crescimento & desenvolvimento , Células Cultivadas , Transferência Ressonante de Energia de Fluorescência , Humanos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Fagossomos/microbiologia
14.
Genome Biol Evol ; 8(2): 387-402, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26748339

RESUMO

In mycobacteria, various type VII secretion systems corresponding to different ESX (ESAT-6 secretory) types, are contributing to pathogenicity, iron acquisition, and/or conjugation. In addition to the known chromosomal ESX loci, the existence of plasmid-encoded ESX systems was recently reported. To investigate the potential role of ESX-encoding plasmids on mycobacterial evolution, we analyzed a large representative collection of mycobacterial genomes, including both chromosomal and plasmid-borne sequences. Data obtained for chromosomal ESX loci confirmed the previous five classical ESX types and identified a novel mycobacterial ESX-4-like type, termed ESX-4-bis. Moreover, analysis of the plasmid-encoded ESX loci showed extensive diversification, with at least seven new ESX profiles, identified. Three of them (ESX-P clusters 1-3) were found in multiple plasmids, while four corresponded to singletons. Our phylogenetic and gene-order-analyses revealed two main groups of ESX types: 1) ancestral types, including ESX-4 and ESX-4-like systems from mycobacterial and non-mycobacterial actinobacteria and 2) mycobacteria-specific ESX systems, including ESX-1-2-3-5 systems and the plasmid-encoded ESX types. Synteny analysis revealed that ESX-P systems are part of phylogenetic groups that derived from a common ancestor, which diversified and resulted in the different ESX types through extensive gene rearrangements. A converging body of evidence, derived from composition bias-, phylogenetic-, and synteny analyses points to a scenario in which ESX-encoding plasmids have been a major driving force for acquisition and diversification of type VII systems in mycobacteria, which likely played (and possibly still play) important roles in the adaptation to new environments and hosts during evolution of mycobacterial pathogenesis.


Assuntos
Evolução Molecular , Genoma Bacteriano , Mycobacterium/genética , Plasmídeos/genética , Sistemas de Secreção Tipo IV/genética , Rearranjo Gênico , Transferência Genética Horizontal , Mycobacterium/classificação , Filogenia , Sintenia
15.
Infect Immun ; 83(2): 780-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25486995

RESUMO

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium involved in pulmonary and cutaneo-mucous infections worldwide, to which cystic fibrosis patients are exquisitely susceptible. The analysis of the genome sequence of M. abscessus showed that this bacterium is endowed with the metabolic pathways typically found in environmental microorganisms that come into contact with soil, plants, and aquatic environments, where free-living amoebae are frequently present. M. abscessus also contains several genes that are characteristically found only in pathogenic bacteria. One of them is MAB_0555, encoding a putative phospholipase C (PLC) that is absent from most other rapidly growing mycobacteria, including Mycobacterium chelonae and Mycobacterium smegmatis. Here, we report that purified recombinant M. abscessus PLC is highly cytotoxic to mouse macrophages, presumably due to hydrolysis of membrane phospholipids. We further showed by constructing and using an M. abscessus PLC knockout mutant that loss of PLC activity is deleterious to M. abscessus intracellular survival in amoebae. The importance of PLC is further supported by the fact that M. abscessus PLC was found to be expressed only in amoebae. Aerosol challenge of mice with M. abscessus strains that were precultured in amoebae enhanced M. abscessus lung infectivity relative to M. abscessus grown in broth culture. Our study underlines the importance of PLC for the virulence of M. abscessus. Despite the difficulties of isolating M. abscessus from environmental sources, our findings suggest that M. abscessus has evolved in close contact with environmental protozoa, which supports the argument that amoebae may contribute to the virulence of opportunistic mycobacteria.


Assuntos
Amoeba/fisiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium/patogenicidade , Fosfolipases Tipo C/fisiologia , Amoeba/microbiologia , Animais , Sequência de Bases , Células Cultivadas , Técnicas de Cocultura , Fibrose Cística/microbiologia , Técnicas de Inativação de Genes , Genoma Bacteriano/genética , Macrófagos/imunologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/enzimologia , Mycobacterium/genética , Infecções por Mycobacterium não Tuberculosas/microbiologia , Proteínas Recombinantes , Análise de Sequência de DNA , Fosfolipases Tipo C/genética , Fatores de Virulência/genética
16.
Vet Immunol Immunopathol ; 145(3-4): 611-7, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22297149

RESUMO

Human amebiasis is caused by the protozoan Entamoeba histolytica. This protozoan is responsible for muco-hemorrhagic diarrhoea and liver abscess in affected populations. E. histolytica can be asymptomatic commensally confined to the intestinal lumen or can result in invasion of the colonic mucosa leading to ulceration and/or liver abscesses. Recently, human colonic explants have been identified as valuable in the study of host-parasite interactions. Here we investigated the potential of porcine colonic explants as an alternative to human tissues which are far less available. Porcine colonic explants were cultured with two strains of E. histolytica, one virulent (HM1:IMSS) and one avirulent (Rahman). Results from histopathological and real-time PCR analysis showed that porcine explants cultured with virulent ameba trophozoites react similarly to their human counterparts with an invasion of the tissue by the trophozoites and the triggering of typical innate immune response against the parasite. On the contrary, explants cultured with avirulent ameba trophozoites were preserved. The study open the way to the use of porcine colonic explants in the study of the complex interactions between the parasite and the host.


Assuntos
Colo/parasitologia , Entamoeba histolytica/imunologia , Interações Hospedeiro-Parasita/imunologia , Animais , Quimiocinas/genética , Colo/imunologia , Humanos , Imunidade Inata , RNA Mensageiro/análise , Suínos
17.
PLoS One ; 6(12): e28795, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22205970

RESUMO

BACKGROUND: Entamoeba histolytica is an important parasite of the human intestine. Its life cycle is monoxenous with two stages: (i) the trophozoite, growing in the intestine and (ii) the cyst corresponding to the dissemination stage. The trophozoite in the intestine can live as a commensal leading to asymptomatic infection or as a tissue invasive form producing mucosal ulcers and liver abscesses. There is no animal model mimicking the whole disease cycle. Most of the biological information on E. histolytica has been obtained from trophozoite adapted to axenic culture. The reproduction of intestinal amebiasis in an animal model is difficult while for liver amebiasis there are well-described rodent models. During this study, we worked on the assessment of pigs as a new potential model to study amebiasis. METHODOLOGY/PRINCIPAL FINDINGS: We first co-cultured trophozoites of E. histolytica with porcine colonic fragments and observed a disruption of the mucosal architecture. Then, we showed that outbred pigs can be used to reproduce some lesions associated with human amebiasis. A detailed analysis was performed using a washed closed-jejunal loops model. In loops inoculated with virulent amebas a severe acute ulcerative jejunitis was observed with large hemorrhagic lesions 14 days post-inoculation associated with the presence of the trophozoites in the depth of the mucosa in two out four animals. Furthermore, typical large sized hepatic abscesses were observed in the liver of one animal 7 days post-injection in the portal vein and the liver parenchyma. CONCLUSIONS: The pig model could help with simultaneously studying intestinal and extraintestinal lesion development.


Assuntos
Modelos Animais de Doenças , Disenteria Amebiana , Suínos , Animais , Técnicas de Cocultura , Colo/citologia , Colo/parasitologia , Disenteria Amebiana/parasitologia , Entamoeba histolytica/crescimento & desenvolvimento , Entamoeba histolytica/patogenicidade , Feminino , Humanos , Injeções , Jejuno/citologia , Jejuno/parasitologia , Abscesso Hepático Amebiano/parasitologia , Veia Porta/parasitologia , Fatores de Tempo , Trofozoítos/fisiologia
18.
Dev Comp Immunol ; 34(5): 562-71, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20045027

RESUMO

Suppressor of cytokine signaling (SOCS) proteins are key physiological regulators of both innate and adaptive immunity. These proteins belong to the three major classes of modulators of cytokines signaling. In the following article, we used porcine polarized intestinal cells to study early response to the protozoan, Entamoeba histolytica, and we identified by rapid amplification of cDNA ends (RACE) PCR porcine SOCS1, SOCS4, SOCS5 and SOCS6 encoding sequences. With more than 92% identity predicted porcine SOCS proteins are very similar to their human counterparts. Among SOCS transcripts, only SOCS2 mRNA was significantly induced in epithelial intestinal cells in response to the cytolytic activity of the parasite. The transcriptomic profile obtained after 3h of co-culture of polarized intestinal cells with E. histolytica was clearly oriented toward inflammation and the recruitment of neutrophils. These transcriptomic data have been normalized with accuracy by the utilisation of multiple validated reference genes. The analysis offers a first set of reference genes useful for future studies in porcine intestinal cells. Our data shed light on the understanding of the early response of polarized intestinal cells to E. histolytica and identified a potential involvement of SOCS2 in the parasite regulation of the host response.


Assuntos
Entamoeba histolytica/imunologia , Entamebíase/imunologia , Mucosa Intestinal/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Sequência de Aminoácidos , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Entamoeba histolytica/patogenicidade , Entamebíase/genética , Entamebíase/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Parasita , Humanos , Inflamação , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Dados de Sequência Molecular , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Homologia de Sequência de Aminoácidos , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/imunologia , Suínos
19.
Mol Immunol ; 46(5): 927-36, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19110312

RESUMO

Amoebiasis caused by Entamoebahistolytica triggers an acute inflammatory response at early stages of intestinal infection. The patho-physiological study of intestinal amoebiasis requires the development of powerful animal models. Swine provide robust model for human diseases and they could be used to study intestinal amoebiasis. Here, we introduce an in vitro model of swine intestinal epithelial cell (IPI-2I) co-cultured with E. histolytica. Intestinal epithelial cells (IECs) have crucial roles in sensing pathogens and initiating innate immune response, which qualitatively influence adaptive immune response against them. The contact between the two cells induces marked macroscopic lesions of IEC monolayer and striking alteration of the IPI-2I cell phenotype including blebbing, such as loss of attachment before to be phagocyte by the trophozoite. Increase in Lactate Dehydrogenase (LDH) levels in the culture supernatant of IECs was observed when ameba is present and could reflect the cellular cytotoxicity exerted by the parasite. Using quantitative real-time PCR, we identified the up-regulation of cytokines/chemokines implicated in neutrophil chemoattraction and inflammation, such as CCL2, CCL20, CXCL2, CXCL3, GM-CSF, IL1 alpha, IL6 and IL8, in response to the parasite that can further regulate the immunoregulatory functions of the immune cells of the host. The study points a cardinal role of these pro-inflammatory compounds as central mediators in the interaction IECs/ameba and suggests mechanisms by which they coordinate intestinal immune response. This will focus future efforts on delineating the molecular and cellular mechanisms of other cell partners by the way of in vivo infection of swine.


Assuntos
Disenteria Amebiana/imunologia , Disenteria Amebiana/veterinária , Entamoeba histolytica/imunologia , Mucosa Intestinal/imunologia , Jejuno/imunologia , Doenças dos Suínos/imunologia , Suínos/imunologia , Animais , Linhagem Celular , Citocinas/imunologia , Disenteria Amebiana/parasitologia , Imunidade Inata/imunologia , Mucosa Intestinal/parasitologia , Jejuno/parasitologia , Suínos/parasitologia , Suínos/psicologia , Doenças dos Suínos/parasitologia , Regulação para Cima/imunologia
20.
Mol Biochem Parasitol ; 157(2): 236-40, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18160113

RESUMO

Calreticulin (CRT), an intracellular chaperone protein, is crucial for proper folding and transport of proteins through the endoplasmic reticulum (ER). It has recently been identified as a critical regulator of some several different cellular functions such as migration, phagocytosis of apoptotic cells and cytotoxic T lymphocyte- or natural killer cell-mediated lysis. Characterization of CRT isolated from parasites may thus help to decipher the contribution of this protein in the parasites' biology and host-parasite interactions. Here, we report descriptive data on the localization of Entamoeba histolytica's CRT at rest and following cap formation by Concanavalin A. As expected, CRT from E. histolytica localizes in the ER. However, the protein was surprisingly found to localize to the parasite surface and, furthermore, to concentrate in the uropod following activation of surface receptors by capping with Concanavalin A.


Assuntos
Calreticulina/análise , Retículo Endoplasmático/química , Entamoeba histolytica/química , Proteínas de Membrana/análise , Animais , Entamoeba histolytica/citologia , Microscopia de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...